Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 2823, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38561349

ABSTRACT

Dysfunction in fast-spiking parvalbumin interneurons (PV-INs) may represent an early pathophysiological perturbation in Alzheimer's Disease (AD). Defining early proteomic alterations in PV-INs can provide key biological and translationally-relevant insights. We used cell-type-specific in-vivo biotinylation of proteins (CIBOP) coupled with mass spectrometry to obtain native-state PV-IN proteomes. PV-IN proteomic signatures include high metabolic and translational activity, with over-representation of AD-risk and cognitive resilience-related proteins. In bulk proteomes, PV-IN proteins were associated with cognitive decline in humans, and with progressive neuropathology in humans and the 5xFAD mouse model of Aß pathology. PV-IN CIBOP in early stages of Aß pathology revealed signatures of increased mitochondria and metabolism, synaptic and cytoskeletal disruption and decreased mTOR signaling, not apparent in whole-brain proteomes. Furthermore, we demonstrated pre-synaptic defects in PV-to-excitatory neurotransmission, validating our proteomic findings. Overall, in this study we present native-state proteomes of PV-INs, revealing molecular insights into their unique roles in cognitive resiliency and AD pathogenesis.


Subject(s)
Alzheimer Disease , Mice , Humans , Animals , Alzheimer Disease/metabolism , Parvalbumins/metabolism , Proteomics , Proteome/metabolism , Interneurons/metabolism , Mice, Transgenic
2.
Cell Rep Methods ; 4(1): 100684, 2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38211592

ABSTRACT

The mammalian brain contains a diverse array of cell types, including dozens of neuronal subtypes with distinct anatomical and functional characteristics. The brain leverages these neuron-type specializations to perform diverse circuit operations and thus execute different behaviors properly. Through the use of Cre lines, access to specific neuron types has improved over past decades. Despite their extraordinary utility, development and cross-breeding of Cre lines is time consuming and expensive, presenting a significant barrier to entry for investigators. Furthermore, cell-based therapeutics developed in Cre mice are not clinically translatable. Recently, several adeno-associated virus (AAV) vectors utilizing neuron-type-specific regulatory transcriptional sequences (enhancer-AAVs) were developed that overcome these limitations. Using a publicly available RNA sequencing (RNA-seq) dataset, we evaluated the potential of several candidate enhancers for neuron-type-specific targeting in the hippocampus. Here, we demonstrate that a previously identified enhancer-AAV selectively targets dentate granule cells over other excitatory neuron types in the hippocampus of wild-type adult mice.


Subject(s)
Dentate Gyrus , Neurons , Mice , Animals , Dentate Gyrus/physiology , Neurons/physiology , Hippocampus/physiology , Mammals
3.
J Vis Exp ; (196)2023 06 30.
Article in English | MEDLINE | ID: mdl-37458444

ABSTRACT

Understanding normal and aberrant in vivo cell behaviors is necessary to develop clinical interventions to thwart disease initiation and progression. It is therefore critical to optimize imaging approaches that facilitate the observation of cell dynamics in situ, where tissue structure and composition remain unperturbed. The epidermis is the body's outermost barrier, as well as the source of the most prevalent human cancers, namely cutaneous skin carcinomas. The accessibility of skin tissue presents a unique opportunity to monitor epithelial and dermal cell behaviors in intact animals using noninvasive intravital microscopy. Nevertheless, this sophisticated imaging approach has primarily been achieved using upright multiphoton microscopes, which represent a significant barrier for entry for most investigators. This study presents a custom-designed, 3D-printed microscope stage insert suitable for use with inverted confocal microscopes, streamlining the long-term intravital imaging of ear skin in live transgenic mice. We believe this versatile invention, which may be customized to fit the inverted microscope brand and model of choice and adapted to image additional organ systems, will prove invaluable to the greater scientific research community by significantly enhancing the accessibility of intravital microscopy. This technological advancement is critical for bolstering our understanding of live cell dynamics in normal and disease contexts.


Subject(s)
Skin Neoplasms , Skin , Mice , Animals , Humans , Skin/diagnostic imaging , Intravital Microscopy/methods , Epidermis , Mice, Transgenic
4.
Elife ; 112022 11 07.
Article in English | MEDLINE | ID: mdl-36341568

ABSTRACT

Understanding the activity of the mammalian brain requires an integrative knowledge of circuits at distinct scales, ranging from ion channel gating to circuit connectomics. Computational models are regularly employed to understand how multiple parameters contribute synergistically to circuit behavior. However, traditional models of anatomically and biophysically realistic neurons are computationally demanding, especially when scaled to model local circuits. To overcome this limitation, we trained several artificial neural network (ANN) architectures to model the activity of realistic multicompartmental cortical neurons. We identified an ANN architecture that accurately predicted subthreshold activity and action potential firing. The ANN could correctly generalize to previously unobserved synaptic input, including in models containing nonlinear dendritic properties. When scaled, processing times were orders of magnitude faster compared with traditional approaches, allowing for rapid parameter-space mapping in a circuit model of Rett syndrome. Thus, we present a novel ANN approach allowing for rapid, detailed network experiments using inexpensive and commonly available computational resources.


Subject(s)
Models, Neurological , Neocortex , Animals , Neocortex/physiology , Neurons/physiology , Action Potentials/physiology , Computer Simulation , Mammals
5.
Elife ; 112022 06 21.
Article in English | MEDLINE | ID: mdl-35727131

ABSTRACT

In Alzheimer's disease (AD), a multitude of genetic risk factors and early biomarkers are known. Nevertheless, the causal factors responsible for initiating cognitive decline in AD remain controversial. Toxic plaques and tangles correlate with progressive neuropathology, yet disruptions in circuit activity emerge before their deposition in AD models and patients. Parvalbumin (PV) interneurons are potential candidates for dysregulating cortical excitability as they display altered action potential (AP) firing before neighboring excitatory neurons in prodromal AD. Here, we report a novel mechanism responsible for PV hypoexcitability in young adult familial AD mice. We found that biophysical modulation of Kv3 channels, but not changes in their mRNA or protein expression, were responsible for dampened excitability in young 5xFAD mice. These K+ conductances could efficiently regulate near-threshold AP firing, resulting in gamma-frequency-specific network hyperexcitability. Thus, biophysical ion channel alterations alone may reshape cortical network activity prior to changes in their expression levels. Our findings demonstrate an opportunity to design a novel class of targeted therapies to ameliorate cortical circuit hyperexcitability in early AD.


Subject(s)
Alzheimer Disease , Parvalbumins , Shaw Potassium Channels/metabolism , Action Potentials/physiology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Biophysical Phenomena , Interneurons/physiology , Mice , Neurons/metabolism , Parvalbumins/metabolism
6.
Brain Neurosci Adv ; 6: 23982128221086464, 2022.
Article in English | MEDLINE | ID: mdl-35359460

ABSTRACT

Synapse loss is associated with cognitive decline in Alzheimer's disease, and owing to their plastic nature, synapses are an ideal target for therapeutic intervention. Oligomeric amyloid beta around amyloid plaques is known to contribute to synapse loss in mouse models and is associated with synapse loss in human Alzheimer's disease brain tissue, but the mechanisms leading from Aß to synapse loss remain unclear. Recent data suggest that the fast-activating and -inactivating voltage-gated potassium channel subtype 3.4 (Kv3.4) may play a role in Aß-mediated neurotoxicity. Here, we tested whether this channel could also be involved in Aß synaptotoxicity. Using adeno-associated virus and clustered regularly interspaced short palindromic repeats technology, we reduced Kv3.4 expression in neurons of the somatosensory cortex of APP/PS1 mice. These mice express human familial Alzheimer's disease-associated mutations in amyloid precursor protein and presenilin-1 and develop amyloid plaques and plaque-associated synapse loss similar to that observed in Alzheimer's disease brain. We observe that reducing Kv3.4 levels ameliorates dendritic spine loss and changes spine morphology compared to control virus. In support of translational relevance, Kv3.4 protein was observed in human Alzheimer's disease and control brain and is associated with synapses in human induced pluripotent stem cell-derived cortical neurons. We also noted morphological changes in induced pluripotent stem cell neurones challenged with human Alzheimer's disease-derived brain homogenate containing Aß but, in this in vitro model, total mRNA levels of Kv3.4 were found to be reduced, perhaps as an early compensatory mechanism for Aß-induced damage. Overall, our results suggest that approaches to reduce Kv3.4 expression and/or function in the Alzheimer's disease brain could be protective against Aß-induced synaptic alterations.

7.
J Neurosci ; 41(31): 6596-6616, 2021 08 04.
Article in English | MEDLINE | ID: mdl-34261699

ABSTRACT

Eukaryotic cells maintain proteostasis through mechanisms that require cytoplasmic and mitochondrial translation. Genetic defects affecting cytoplasmic translation perturb synapse development, neurotransmission, and are causative of neurodevelopmental disorders, such as Fragile X syndrome. In contrast, there is little indication that mitochondrial proteostasis, either in the form of mitochondrial protein translation and/or degradation, is required for synapse development and function. Here we focus on two genes deleted in a recurrent copy number variation causing neurodevelopmental disorders, the 22q11.2 microdeletion syndrome. We demonstrate that SLC25A1 and MRPL40, two genes present in the microdeleted segment and whose products localize to mitochondria, interact and are necessary for mitochondrial ribosomal integrity and proteostasis. Our Drosophila studies show that mitochondrial ribosome function is necessary for synapse neurodevelopment, function, and behavior. We propose that mitochondrial proteostasis perturbations, either by genetic or environmental factors, are a pathogenic mechanism for neurodevelopmental disorders.SIGNIFICANCE STATEMENT The balance between cytoplasmic protein synthesis and degradation, or cytoplasmic proteostasis, is required for normal synapse function and neurodevelopment. Cytoplasmic and mitochondrial ribosomes are necessary for two compartmentalized, yet interdependent, forms of proteostasis. Proteostasis dependent on cytoplasmic ribosomes is a well-established target of genetic defects that cause neurodevelopmental disorders, such as autism. Here we show that the mitochondrial ribosome is a neurodevelopmentally regulated organelle whose function is required for synapse development and function. We propose that defective mitochondrial proteostasis is a mechanism with the potential to contribute to neurodevelopmental disease.


Subject(s)
Developmental Disabilities , Mitochondria/physiology , Mitochondrial Proteins/genetics , Organic Anion Transporters/genetics , Proteostasis/genetics , Ribonucleoproteins/genetics , Ribosomal Proteins/genetics , Animals , Cell Line , Developmental Disabilities/genetics , Developmental Disabilities/metabolism , Developmental Disabilities/physiopathology , Drosophila , Gene Expression Regulation/genetics , Humans , Neurogenesis/physiology , Protein Biosynthesis/genetics , Rats , Rats, Sprague-Dawley , Ribosomes/physiology
8.
Sci Rep ; 11(1): 6065, 2021 03 16.
Article in English | MEDLINE | ID: mdl-33727679

ABSTRACT

A common electrophysiology technique used in neuroscience is patch clamp: a method in which a glass pipette electrode facilitates single cell electrical recordings from neurons. Typically, patch clamp is done manually in which an electrophysiologist views a brain slice under a microscope, visually selects a neuron to patch, and moves the pipette into close proximity to the cell to break through and seal its membrane. While recent advances in the field of patch clamping have enabled partial automation, the task of detecting a healthy neuronal soma in acute brain tissue slices is still a critical step that is commonly done manually, often presenting challenges for novices in electrophysiology. To overcome this obstacle and progress towards full automation of patch clamp, we combined the differential interference microscopy optical technique with an object detection-based convolutional neural network (CNN) to detect healthy neurons in acute slice. Utilizing the YOLOv3 convolutional neural network architecture, we achieved a 98% reduction in training times to 18 min, compared to previously published attempts. We also compared networks trained on unaltered and enhanced images, achieving up to 77% and 72% mean average precision, respectively. This novel, deep learning-based method accomplishes automated neuronal detection in brain slice at 18 frames per second with a small data set of 1138 annotated neurons, rapid training time, and high precision. Lastly, we verified the health of the identified neurons with a patch clamp experiment where the average access resistance was 29.25 M[Formula: see text] (n = 9). The addition of this technology during live-cell imaging for patch clamp experiments can not only improve manual patch clamping by reducing the neuroscience expertise required to select healthy cells, but also help achieve full automation of patch clamping by nominating cells without human assistance.


Subject(s)
Brain/cytology , Deep Learning , Image Processing, Computer-Assisted , Microdissection , Neurons/cytology , Animals , Brain/physiology , Microscopy , Neurons/physiology
9.
Neuron ; 99(5): 999-1015.e6, 2018 09 05.
Article in English | MEDLINE | ID: mdl-30122378

ABSTRACT

Purkinje cell dendrites convert excitatory climbing fiber input into signals that instruct plasticity and motor learning. Modulation of instructive signaling may increase the range in which learning is encoded, yet the mechanisms that allow for this are poorly understood. We found that optogenetic activation of molecular layer interneurons (MLIs) that inhibit Purkinje cells suppressed climbing-fiber-evoked dendritic Ca2+ spiking. Inhibitory suppression of Ca2+ spiking depended on the level of MLI activation and influenced the induction of associative synaptic plasticity, converting climbing-fiber-mediated potentiation of parallel fiber-evoked responses into depression. In awake mice, optogenetic activation of floccular climbing fibers in association with head rotation produced an adaptive increase in the vestibulo-ocular reflex (VOR). However, when climbing fibers were co-activated with MLIs, adaptation occurred in the opposite direction, decreasing the VOR. Thus, MLIs can direct a continuous spectrum of plasticity and learning through their influence on Purkinje cell dendritic Ca2+ signaling.


Subject(s)
Cerebellum/cytology , Cerebellum/physiology , Learning/physiology , Neural Inhibition/physiology , Neuronal Plasticity/physiology , Purkinje Cells/physiology , Animals , Cerebellum/chemistry , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Optogenetics/methods , Purkinje Cells/chemistry
10.
Elife ; 72018 08 17.
Article in English | MEDLINE | ID: mdl-30117806

ABSTRACT

Motor learning involves neural circuit modifications in the cerebellar cortex, likely through re-weighting of parallel fiber inputs onto Purkinje cells (PCs). Climbing fibers instruct these synaptic modifications when they excite PCs in conjunction with parallel fiber activity, a pairing that enhances climbing fiber-evoked Ca2+ signaling in PC dendrites. In vivo, climbing fibers spike continuously, including during movements when parallel fibers are simultaneously conveying sensorimotor information to PCs. Whether parallel fiber activity enhances climbing fiber Ca2+ signaling during motor behaviors is unknown. In mice, we found that inhibitory molecular layer interneurons (MLIs), activated by parallel fibers during practiced movements, suppressed parallel fiber enhancement of climbing fiber Ca2+ signaling in PCs. Similar results were obtained in acute slices for brief parallel fiber stimuli. Interestingly, more prolonged parallel fiber excitation revealed latent supralinear Ca2+ signaling. Therefore, the balance of parallel fiber and MLI input onto PCs regulates concomitant climbing fiber Ca2+ signaling.


Subject(s)
Cerebellar Cortex/physiology , Dendrites/physiology , Movement/physiology , Purkinje Cells/physiology , Animals , Calcium Signaling/physiology , Evoked Potentials, Motor/physiology , Excitatory Postsynaptic Potentials/physiology , Interneurons/physiology , Learning/physiology , Mice , Patch-Clamp Techniques , Synapses/physiology
11.
PLoS One ; 12(6): e0179347, 2017.
Article in English | MEDLINE | ID: mdl-28658323

ABSTRACT

The cerebellar system helps modulate and fine-tune motor action. Purkinje cells (PCs) provide the sole output of the cerebellar cortex, therefore, any cerebellar involvement in motor activity must be driven by changes in PC firing rates. Several different cell types influence PC activity including excitatory input from parallel fibers and inhibition from molecular layer interneurons (MLIs). Similar to PCs, MLI activity is driven by parallel fibers, therefore, MLIs provide feed-forward inhibition onto PCs. To aid in the experimental assessment of how molecular layer inhibition contributes to cerebellar function and motor behavior, we characterized a new knock-in mouse line with Cre recombinase expression under control of endogenous c-kit transcriptional machinery. Using these engineered c-Kit mice, we were able to obtain high levels of conditional MLI transduction in adult mice using Cre-dependent viral vectors without any PC or granule cell labeling. We then used the mouse line to target MLIs for activity perturbation in vitro using opto- and chemogenetics.


Subject(s)
Cerebellar Cortex/cytology , Cerebellum/cytology , Interneurons/cytology , Proto-Oncogene Proteins c-kit/metabolism , Action Potentials/physiology , Animals , Cerebellar Cortex/metabolism , Cerebellum/metabolism , Interneurons/metabolism , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-kit/genetics
12.
Cell Rep ; 18(8): 2018-2029, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28228266

ABSTRACT

In many neurons, subthreshold depolarization in the soma can transiently increase action-potential (AP)-evoked neurotransmission via analog-to-digital facilitation. The mechanisms underlying this form of short-term synaptic plasticity are unclear, in part, due to the relative inaccessibility of the axon to direct physiological interrogation. Using voltage imaging and patch-clamp recording from presynaptic boutons of cerebellar stellate interneurons, we observed that depolarizing somatic potentials readily spread into the axon, resulting in AP broadening, increased spike-evoked Ca2+ entry, and enhanced neurotransmission strength. Kv3 channels, which drive AP repolarization, rapidly inactivated upon incorporation of Kv3.4 subunits. This leads to fast susceptibility to depolarization-induced spike broadening and analog facilitation independent of Ca2+-dependent protein kinase C signaling. The spread of depolarization into the axon was attenuated by hyperpolarization-activated currents (Ih currents) in the maturing cerebellum, precluding analog facilitation. These results suggest that analog-to-digital facilitation is tempered by development or experience in stellate cells.


Subject(s)
Action Potentials/physiology , Presynaptic Terminals/metabolism , Shaw Potassium Channels/metabolism , Signal Transduction/physiology , Synapses/metabolism , Synapses/physiology , Animals , Axons/metabolism , Axons/physiology , Calcium/metabolism , Cerebellum/metabolism , Cerebellum/physiology , Interneurons/metabolism , Interneurons/physiology , Mice , Neurons/metabolism , Neurons/physiology , Patch-Clamp Techniques , Potassium Channels/metabolism , Presynaptic Terminals/physiology , Protein Kinase C/metabolism , Synaptic Transmission/physiology
13.
Neuron ; 91(2): 370-83, 2016 07 20.
Article in English | MEDLINE | ID: mdl-27346528

ABSTRACT

In axons, an action potential (AP) is thought to be broadcast as an unwavering binary pulse over its arbor, driving neurotransmission uniformly at release sites. Yet by recording from axons of cerebellar stellate cell (SC) interneurons, we show that AP width varies between presynaptic bouton sites, even within the same axon branch. The varicose geometry of SC boutons alone does not impose differences in spike duration. Rather, axonal patching revealed heterogeneous peak conductance densities of currents mediated mainly by fast-activating Kv3-type potassium channels, with clustered hotspots at boutons and restricted expression at adjoining shafts. Blockade of Kv channels at individual boutons indicates that currents immediately local to a release site direct spike repolarization at that location. Thus, the clustered arrangement and variable expression density of Kv3 channels at boutons are key determinants underlying compartmentalized control of AP width in a near synapse-by-synapse manner, multiplying the signaling capacity of these structures.


Subject(s)
Action Potentials/physiology , Axons/physiology , Potassium Channels/physiology , Presynaptic Terminals/physiology , Synapses/physiology , Animals , Cerebellum/physiology , Interneurons/physiology , Mice, Inbred C57BL , Patch-Clamp Techniques/methods , Synaptic Transmission/physiology
14.
J Neurosci ; 34(19): 6611-23, 2014 May 07.
Article in English | MEDLINE | ID: mdl-24806686

ABSTRACT

The discrete arrangement of voltage-gated K(+) (Kv) channels in axons may impart functional advantages in action potential (AP) signaling yet, in compact cell types, the organization of Kv channels is poorly understood. We find that in cerebellar stellate cell interneurons of mice, the composition and influence of Kv channels populating the axon is diverse and depends on location allowing axonal compartments to differentially control APs in a local manner. Kv1 channels determine AP repolarization at the spike initiation site but not at more distal sites, limiting the expression of use-dependent spike broadening to the most proximal axon region, likely a key attribute informing spiking phenotype. Local control of AP repolarization at presynaptic boutons depends on Kv3 channels keeping APs brief, thus limiting Ca(2+) influx and synaptic strength. These observations suggest that AP repolarization is tuned by the local influence of distinct Kv channel types, and this organization enhances the functional segregation of axonal compartments.


Subject(s)
Axons/physiology , Cerebellum/physiology , Interneurons/physiology , Potassium Channels/physiology , Presynaptic Terminals/physiology , Receptors, Presynaptic/physiology , Animals , Cerebellum/cytology , Electrophysiological Phenomena , Female , Fluorescent Dyes , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Patch-Clamp Techniques , Receptors, AMPA/physiology , Receptors, GABA-A/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Shaker Superfamily of Potassium Channels/physiology , Shaw Potassium Channels/physiology , Synaptic Transmission/physiology
15.
Adv Exp Med Biol ; 775: 85-99, 2013.
Article in English | MEDLINE | ID: mdl-23392926

ABSTRACT

Taurine activates not only Cl(-)-permeable ionotropic receptors but also receptors that mediate metabotropic responses. The metabotropic property of taurine was revealed in electrophysiological recordings obtained after fully blocking Cl(-)-permeable receptors with an inhibitory "cocktail" consisting of picrotoxin, SR95531, and strychnine. We found that taurine's metabotropic effects regulate voltage-gated channels in retinal neurons. After applying the inhibitory cocktail, taurine enhanced delayed outward rectifier K(+) channels preferentially in Off-bipolar cells, and the effect was completely blocked by the specific PKC inhibitor, GF109203X. Additionally, taurine also acted through a metabotropic pathway to suppress both L- and N-type Ca(2+) channels in retinal neurons, which were insensitive to the potent GABA(B) receptor inhibitor, CGP55845. This study reinforces our previous finding that taurine in physiological concentrations produces a multiplicity of metabotropic effects that precisely govern the integration of signals being transmitted from the retina to the brain.


Subject(s)
Calcium Channels/metabolism , Potassium Channels, Voltage-Gated/metabolism , Retinal Neurons/drug effects , Retinal Neurons/metabolism , Taurine/pharmacology , Ambystoma/metabolism , Animals , Calcium Channels, L-Type/metabolism , Calcium Channels, N-Type/metabolism , Protein Kinase C , Receptors, Metabotropic Glutamate/metabolism , Retinal Bipolar Cells/cytology , Retinal Bipolar Cells/drug effects , Retinal Bipolar Cells/metabolism , Retinal Neurons/cytology , Signal Transduction/drug effects
16.
J Physiol ; 588(Pt 20): 3943-56, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20807794

ABSTRACT

Excitatory amino acid transporters (EAATs) are responsible for extracellular glutamate uptake within the retina, and are expressed by retinal neurons and Müller cells. Their role within glutamatergic synapses is not completely understood. In the salamander retina, five distinct EAAT-encoding genes have been cloned, making the amphibian retina an excellent system to study EAAT function. This study focused on sEAAT2, which is expressed in photoreceptor terminals and Off-bipolar cells in two isoforms, sEAAT2A and sEAAT2B. Using whole-cell patch-clamp recording, florescence imaging and antibody labelling methods, we systematically studied the functions of these two isoforms at the synapse between photoreceptors and bipolar cells, both in dark and with photic stimulation. Both sEAAT2A and sEAAT2B were sensitive to dihydrokainic acid (DHKA), a known EAAT2-specific inhibitor. Each isoform of sEAAT2 was found to play a role in tonic glutamate uptake at the cone synapse in darkness. Furthermore, presynaptic sEAAT2A strongly suppressed the rapid, transient glutamate signal from cones following light-offset. This was achieved by quickly binding exocytosed glutamate, which subsequently limited glutamate spillover to adjacent receptors at postsynaptic sites. Since the intensity and duration of photic stimulation determine the magnitude of these cone transient signals, we postulate that presynaptic cone EAATs contribute to the encoding of contrast sensitivity in cone vision.


Subject(s)
Excitatory Amino Acid Transporter 2/metabolism , Glutamic Acid/metabolism , Retinal Bipolar Cells/physiology , Retinal Cone Photoreceptor Cells/physiology , Vision, Ocular/physiology , Animals , Immunohistochemistry , Light , Patch-Clamp Techniques , Photic Stimulation , Synapses/metabolism , Synaptic Transmission/physiology , Urodela
SELECTION OF CITATIONS
SEARCH DETAIL
...